EP2300011A1 - Procédés et composés thérapeutiques - Google Patents

Procédés et composés thérapeutiques

Info

Publication number
EP2300011A1
EP2300011A1 EP09810870A EP09810870A EP2300011A1 EP 2300011 A1 EP2300011 A1 EP 2300011A1 EP 09810870 A EP09810870 A EP 09810870A EP 09810870 A EP09810870 A EP 09810870A EP 2300011 A1 EP2300011 A1 EP 2300011A1
Authority
EP
European Patent Office
Prior art keywords
cancer
diketopiperazine
pharmaceutically
acid
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP09810870A
Other languages
German (de)
English (en)
Other versions
EP2300011A4 (fr
Inventor
David Bar-Or
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ampio Pharmaceuticals Inc
Original Assignee
DMI Life Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by DMI Life Sciences Inc filed Critical DMI Life Sciences Inc
Publication of EP2300011A1 publication Critical patent/EP2300011A1/fr
Publication of EP2300011A4 publication Critical patent/EP2300011A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/06Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members
    • C07D241/08Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members with oxygen atoms directly attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • This invention relates to diketopiperazines and pharmaceutical compositions comprising them as the active ingredient.
  • the invention also relates to the therapeutic treatments that utilize the diketopiperazines, including inhibition of a proliferative disease or condition, inhibition of angiogenesis, treatment of an angiogenic disease or condition, treatment of cancer and precancerous conditions, treatment of a fibrotic disorder, treatment of a viral infection, inhibition of Akt activation, treatment of an Akt-mediated disease or condition, and inhibition of the production, release or both of matrix metalloproteinase-9.
  • R 1 is:
  • amino acid is glycine, alanine, valine, norvaline, ⁇ -aminoisobutyric acid, 2,4-diaminobutyric acid, 2,3- diaminobutyric acid, leucine, isoleucine, norleucine, serine, homoserine, threonine, aspartic acid, asparagine, glutamic acid, glutamine, lysine, hydroxylysine, histidine, arginine, homoarginine, citrulline, phenylalanine,
  • R 3 may independently be a substituted or unsubstituted alkyl, cycloalkyl, heterocycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl; (iii) a -COOH group replaced by a -COOR 3 group, wherein each R 3 may independently be a substituted or unsubstituted alkyl, cycloalkyl, heterocycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl; (iv) a -COOH group replaced by a -CON(R 4 ) 2 group, wherein each R 4 may independently be H or a substituted or unsubstituted alkyl, cycloalkyl, heterocycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl; (v) an -SH group replaced by -S-S-CH 2 -CH(NH 2 )-COOH or -S-S
  • R 2 has formula ⁇ , HI or IV:
  • each R 5 is independently aryl, heteroaryl, alkyl, cycloalkyl, heterocycloalkyl, alkoxy, aryloxy, acyl, carboxyl, hydroxyl, halogen, amino, nitro, sulfo or sulfhydryl, wherein each alkyl is optionally substituted with hydroxyl, amino or sulfhydryl; n is from O to 5; and
  • R 6 is hydrogen or lower alkyl.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and an active ingredient, wherein the active ingredient is a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention further provides a method of treating a proliferative disease or condition.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention provides a method of inhibiting angiogenesis.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention further provides a method of treating an angiogenic disease or condition.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention provides a method of treating a cancer.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention further provides a method of treating a precancerous condition.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention also provides a method of treating a fibrotic disorder.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention provides a method of treating a viral infection.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention also provides a method of inhibiting the production, release or both of matrix metalloproteinase-9 (MMP-9) by cells.
  • the method comprises contacting the cells with an effective amount of a diketopiperazine of formula I or a pharmaceutically- acceptable salt or prodrug thereof.
  • the invention provides a method of inhibiting the activation
  • the method comprises contacting the cells with an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the invention further provides a method of treating an Akt-mediated disease or condition.
  • the method comprises administering to an animal in need thereof an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • Figure 1 Schematic illustrating the synthesis of biphenyl-4-yl-(3,6-dioxo- piperazin-2-yl)-acetic acid methyl ester (Cpd. 5).
  • RMs (a) a side chain of an amino acid, wherein the amino acid is glycine, alanine, valine, norvaline, ⁇ -aminoisobutyric acid, 2,4-diaminobutyric acid, 2,3- diaminobutyric acid, leucine, isoleucine, norleucine, serine, homoserine, threonine, aspartic acid, asparagine, glutamic acid, glutamine, lysine, hydroxylysine, histidine, arginine, homoarginine, citrulline, phenylalanine,
  • (C) a derivative of a side chain of an amino acid, wherein the amino acid is one of those recited in (a), and the derivatized side chain has:
  • each R 3 may independently be a substituted or unsubstituted alkyl, cycloalkyl, heterocycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl; ( ⁇ ) an -OH group replaced by an -0-PO 3 H 2 or -OR 3 group, wherein each R 3 may independently be a substituted or unsubstituted alkyl, cycloalkyl, heterocycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl; (hi) a -COOH group replaced by a -COOR 3 group, wherein each R 3 may independently be a substituted or unsubstituted alkyl, cycloalkyl, heterocycloalkyl, aryl, alkylaryl, arylalkyl or heteroaryl; (iv) a -NH 2 group replaced by an -NHR 3 or -N(R 3 ) 2 group, wherein each R 3 may independently
  • R 2 has formula ⁇ , DI or IV:
  • each R 5 is independently aryl, heteroaryl, alkyl, cycloalkyl, heterocycloalkyl, alkoxy, aryloxy, acyl, carboxyl, hydroxyl, halogen, amino, nitro, sulfo or sylfhydryl, wherein each alkyl is optionally substituted with hydroxyl, amino or sulfhydryl; n is from 0 to 5; and R 6 is hydrogen or lower alkyl.
  • R 1 is a side chain of glycine, alanine, valine, norvaline, ⁇ - aminoisobutyric acid, 2,4-diaminobutyric acid, 2,3-diaminobutyric acid, leucine, isoleucine, norleucine, serine, homoserine, threonine, aspartic acid, asparagine, glutamic acid, glutamine, lysine, hydroxylysine, histidine, arginine, homoarginine, citrulline, phenylalanine, p-aminophenylalanine, tyrosine, tryptophan, thyroxine or ornithine, or is a derivative of one of these side chains.
  • R 1 is a side chain of glycine, alanine, valine, norvaline, ⁇ - aminoisobutyric acid, 2,4-diaminobutyric acid, 2,3-diaminobutyric acid, leucine, isoleucine, norleucine, serine, homoserine, threonine, aspartic acid, asparagine, glutamic acid, glutamine, lysine, hydroxylysine, arginine, homoarginine, citrulline or ornithine, or is a derivative of one of these chains.
  • R 1 is the side chain of glycine, alanine, valine, leucine or isoleucine, more preferably glycine or alanine, most preferably glycine.
  • R 2 has formula ⁇ or EI, most preferably ⁇ .
  • R 5 is preferably aryl, heteroaryl or aryloxy. More preferably R 5 is aryl or aryloxy.
  • R 5 is phenyl or phenoxy.
  • n is 1-3. Most preferably n is 1.
  • R 5 is preferably in the 4 (para) position on the ring.
  • R 6 is preferably methyl.
  • the most highly preferred compound is biphenyl-4-yl-(3,6-dioxo-piperazin-2-yl)- acetic acid methyl ester (referred to as Cpd. 5 herein).
  • replacement is meant that, with reference to the formula of an amino acid side chain, the specified group is replaced by the other specified group.
  • the formula of the isoleucine side chain is -CH(CH 3 )-CH 2 -CH 3 . If the terminal -CH 3 group is replaced with a
  • side chain of an amino acid is meant that portion of the amino acid attached to
  • the side chain of glycine is -H
  • the side chain of alanine is -CH 3
  • the side chain of serine is -CH 2 OH.
  • acyl is meant a moiety of the formula -C(O)R 7 , wherein R 7 is hydrogen, alkyl, cycloalkyl or aryl.
  • alkoxy is meant a moiety of the formula -OR 8 , wherein R 8 is alkyl.
  • An example of an alkoxy group is methoxy (-0-CH 3 ).
  • alkyl is meant a monovalent saturated straight-chain or branched hydrocarbon containing 1-10 carbon atoms, preferably 1-8, carbon atoms. Each alkyl may, optionally, be substituted with one or more amino, hydroxyl or sulfhydryl groups.
  • “Lower alkyl” means a monovalent saturated straight-chain or branched hydrocarbon containing 1-6 carbon atoms.
  • alkylaryl is meant a lower alkyl having an H replaced by an aryl (e.g.,
  • amino is meant a moiety of the formula -NR 9 R 10 , wherein each R 9 and R 10 is independently H or lower alkyl.
  • aryl is meant a monovalent mono-, bi- or tricyclic aromatic hydrocarbon moiety having 6-14 ring carbon atoms. Preferred is phenyl.
  • aryloxy is meant a moiety of the formula -OR 11 wherein R 11 is an aryl. Preferred is phenoxy.
  • arylalkyl is meant an aryl having an H replaced by a lower alkyl ⁇ e.g., -C 6 H 4 - CH 3 ).
  • cycloalkyl is meant a saturated monovalent mono- or bicyclic hydrocarbon moiety having three to ten ring carbon atoms. Preferably, the cycloalkyl contains 4-8 ring carbon atoms. The most preferred cycloalkyl is cyclohexyl.
  • halogen is meant chlorine, fluorine, bromine or iodine. Preferred is chlorine or bromine.
  • heteroaryl is meant an aryl having at least one, preferably no more than three, of the ring carbon atoms replaced by an O, S or N.
  • heterocycloalkyl is meant a cycloalkyl having at least one, preferably no more than three, of the ring carbon atoms replaced by an O, S or N.
  • hydroxyl is meant -OH.
  • nitro is meant -NO 2 .
  • substituted is meant that the moiety is substituted with one or more substituents selected from the following group: -OH, NH 2 , -SH, -COOH and/or a halogen atom.
  • sulfhydryl is meant -SH.
  • sulfo is meant -SO 3 H or SO 2 .
  • diketopiperazines can be prepared by first synthesizing dipeptides.
  • the dipeptides can be synthesized by methods well known in the art using L-amino acids, D-amino acids or a combination of D- and L-amino acids.
  • dipeptides are also available commercially from numerous sources, including DMI Synthesis Ltd., Cambridge, UK (custom synthesis), Sigma- Aldrich, St. Louis, MO (primarily custom synthesis), Phoenix Pharmaceuticals, Inc.,
  • U.S. Patent Application Publication Number 2004/0024180 describes a method of cyclizing dipeptides. Briefly, the dipeptide is heated in an organic solvent while removing water by distillation.
  • the organic solvent is a low-boiling azeotrope with water, such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n- butanol, 2-butanol, t-butanol, acetic acid butylester, carbon tetrachloride, chlorobenzene chloroform, cyclohexane, 1,2-dichlorethane, diethylacetal, dimethylacetal, acetic acid ethylester, heptane, methylisobutylketone, 3-pentanol, toluene and xylene.
  • water such as acetonitrile, allyl alcohol, benzene, benzyl alcohol, n- butanol, 2-
  • the temperature depends on the reaction speed at which the cyclization takes place and on the type of azeotroping agent used.
  • the reaction is preferably carried out at 50-200 0 C, more preferably 80-15O 0 C.
  • the pH range in which cyclization takes place can be easily determined by the person skilled in the art. It will advantageously be pH 2-9, preferably pH 3-7.
  • the dipeptide is preferably cyclized as described in U.S. Patent No. 6,555,543. Briefly, the dipeptide, with the side-chain carboxyl still protected, is heated under neutral conditions. Typically, the dipeptide will be heated at from about 8O 0 C to about 18O 0 C, preferably at about 12O 0 C.
  • the solvent will be a neutral solvent.
  • the solvent may comprise an alcohol (such as butanol, methanol, ethanol, and higher alcohols, but not phenol) and an azeotropic co-solvent (such as toluene, benzene, or xylene).
  • the alcohol is butan-2-ol
  • the azeotropic co-solvent is toluene.
  • the heating is continued until the reaction is complete, and such times can be determined empirically.
  • the dipeptide will be cyclized by refluxing it for about 8-24 hours, preferably about 18 hours. Finally, the protecting group is removed from the diketopiperazine.
  • strong acids mineral acids, such as sulfuric or hydrochloric acids
  • strong bases alkaline bases, such as potassium hydroxide or sodium hydroxide
  • strong reducing agents ⁇ e.g., lithium aluminum hydride
  • Dipeptides made on solid phase resins can be cyclized and released from the resin in one step. See, e.g., U.S. Patent No. 5,817,751.
  • the resin having an N- alkylated dipeptide attached is suspended in toluene or toluene/ethanol in the presence of acetic acid ⁇ e.g., 1%) or triethylamine ⁇ e.g., 4%).
  • acetic acid ⁇ e.g., 1%) or triethylamine ⁇ e.g., 4%
  • basic cyclization conditions are preferred for their faster cyclization times.
  • amino acid derivatives can be used in the synthesis of the dipeptides, the dipeptides can be derivatized and/or the diketopiperazines can be derivatized, as is known in the art. See, e.g., those references cited above. Also see U.S. Patent No. 5,589,501, U.S. Patent Appl. Pub. No. 2005/0215468, EP Patent Application No. 1,445,323, Chang et al., /. Med. Chem., 16(11): 1277-1280 (1973). The complete disclosures of these references are incorporated herein by reference, as exemplary methods of making amino acid derivatives with R 2 side chains ⁇ e.g., aspartic acid and glutamic acid derivatives).
  • the diketopiperazines of formula I include all possible stereoisomers that can be obtained by varying the configuration of the individual chiral centers, axes or surfaces.
  • the diketopierazines of formula I include all possible diastereomers, as well as all optical isomers (enantiomers).
  • the compound can be synthesized enantioselectively or a mixture of enantiomers and/or diastereomers can be prepared and separated.
  • the resolution of the compounds of the present invention, their starting materials and/or the intermediates may be carried out by known procedures, e.g., as described in the four volume compendium Optical Resolution Procedures for Chemical Compounds: Optical Resolution Information Center, Manhattan College, Riverdale, N. Y., and in Enantiomers, Racemates and Resolutions, Jean Jacques, Andre Collet and Samuel H. Wilen; John Wiley & Sons, Inc., New York, 1981, which are incorporated herein in their entirety.
  • the resolution of the compounds is based on the differences in the physical properties of diastereomers by attachment, either chemically or enzymatically, of an enantiomerically pure moiety, resulting in forms that are separable by fractional crystallization, distillation or chromatography.
  • the pharmaceutically-acceptable salts of the diketopiperazines of formula I may also be used in the practice of the invention.
  • Pharmaceutically-acceptable salts include conventional non-toxic salts, such as salts derived from inorganic acids (such as hydrochloric, hydrobromic, sulfuric, phosphoric, nitric, and the like), organic acids (such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, glutamic, aspartic, benzoic, salicylic, oxalic, ascorbic acid, and the like) or bases (such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation or organic cations derived from N,N-dibenzylethylenediamine, D-glucosamine, or ethylenediamine).
  • the salts are prepared in a conventional manner, e.g., by neutralizing the free base form of the compound with an acid.
  • Prodrug means any compound which releases an active parent drug according to formula I in vivo when such prodrug is administered to an animal.
  • Prodrugs of a diketopiperazine of formula I are prepared by modifying one or more functional group(s) present in the diketopiperazine of formula I in such a way that the modification(s) may be cleaved in vivo to release the parent drug (i.e., the diketopiperazine of formula I).
  • Prodrugs include diketopiperazines of formula I wherein a hydroxy, amino, or sulfhydryl group in a compound of formula I is bonded to any group that may be cleaved in vivo to generate the free hydroxyl, amino, or sulfhydryl group, respectively.
  • Examples of prodrugs include, but are not limited to, esters (e.g., acetate, formate, and benzoate derivatives), carbamates (e.g., N,N-dimethylaminocarbonyl) of hydroxy functional groups in compounds of formula I, and the like.
  • a proliferative disease or condition is a disease or condition causing, caused by, involving, or exacerbated by, proliferation of cells.
  • Specific proliferative diseases and conditions that can be treated with a diketopiperazine of formula I or a pharmaceutically- acceptable salt or prodrug thereof, include blood vessel proliferative disorders, cancer, mesangial cell proliferation disorders, fibrotic disorders and hyperproliferative skin disorders.
  • Blood vessel proliferative disorders include angiogenic diseases and conditions.
  • An angiogenic disease or condition is a disease or condition causing, caused by, involving, exacerbated by, or dependent on angiogenesis.
  • Angiogenesis is the process of new blood vessel formation in the body.
  • Angiogenesis is also used herein to mean the same as, or to include, neovascularization, vascularization, arterialization and vasculogenesis.
  • a diketopiperazine of the invention or a pharmaceutically-acceptable salt or prodrug thereof will inhibit angiogenesis and can be used to treat an angiogenic disease or condition.
  • Specific angiogenic diseases and conditions treatable according to the invention include neoplastic diseases (e.g., tumors (e.g., tumors of the bladder, brain, breast, cervix, colon, rectum, kidney, lung, ovary, pancreas, prostate, stomach and uterus) and tumor metastasis), benign tumors (e.g., hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyrogenic granulomas), hypertrophy (e.g., cardiac hypertrophy induced by thyroid hormone), connective tissue disorders (e.g., arthritis and atherosclerosis), psoriasis, ocular angiogenic diseases (e.g., diabetic retinopathy, retinopathy of prematurity, macular degeneration, cornea
  • the diketopiperazines of the invention will be particularly useful for the treatment of neoplastic diseases and ocular angiogenic diseases (especially diabetic retinopathy and macular degeneration).
  • the diketopiperazines of the invention can also be used to inhibit the vascularization required for embryo implantation, thereby providing a method of birth control.
  • the invention also provides a method of treating a cancer or a precancerous condition.
  • Cancers treatable with a diketopiperazine of formula I, or a pharmaceutically- acceptable salt or prodrug thereof include carcinomas, sarcomas, osteoscarcomas, lymphomas, leukemias, hematologic malignancies, cancer syndromes, malignant tumors, and metastases.
  • Specific cancers treatable according to the invention include brain cancers, head and neck cancers, breast cancers, cardiac cancers, ovarian cancers, cervical cancers, endometrial cancers, urogenital cancers, prostate cancers, gastric cancers, colorectal cancers, pancreatic cancers, bladder cancers, thyroid cancers, hepatic cancers, lung cancers, bone cancers, skin cancers and Kaposi's sarcomas.
  • Specific cancer syndromes treatable according to the invention include Bannayan-Zonana syndrome, Cowden disease and Lhermitte-Duclos disease.
  • malignant tumors treatable according to the invention include malignant tumors of the bladder, bone, brain, breast, cervix, colon, heart, kidney, liver, lung, lymph tissue, ovary, pancreas, prostate, rectum, skin, stomach, thyroid, urogenital and uterus.
  • the diketopiperazines of the invention are especially useful for the treatment of breast cancer and melanoma and for the treatment of metastases.
  • the diketopiperazines of the invention are also especially useful for the treatment of malignant brain tumors, including primary tumors and metastatic (secondary) tumors. About half of all primary brain tumors are gliomas.
  • Gliomas include astrocytomas (e.g., pilocytic astrocytomas, low-grade astrocytomas, anaplastic (high-grade) astrocytomas and glioblastomas multiforme), brain stem gliomas ependymomas, ganglioneuromas, juvenile pilocytic gliomas, mixed gliomas, oligodendrogliomas and optic nerve gliomas.
  • astrocytomas e.g., pilocytic astrocytomas, low-grade astrocytomas, anaplastic (high-grade) astrocytomas and glioblastomas multiforme
  • brain stem gliomas ependymomas ganglioneuromas
  • juvenile pilocytic gliomas mixed gliomas
  • oligodendrogliomas oligodendrogliomas
  • optic nerve gliomas optic nerve gliomas.
  • Metastatic brain tumors are tumors that have spread to the brain from another part of the body. The most common cancers that metastasize to the brain include breast, melanoma and lung cancers. Metastatic brain tumors are the most common form of brain tumor and considerably outnumber primary brain tumors.
  • Precancerous conditions treatable with a diketopiperazine of formula I, or a pharmaceutically-acceptable salt or prodrug thereof include myelodysplastic syndrome, aplastic anemia, cervical lesions, skin nevi (pre-melanoma), prostatic intraepithelial (intraductal) neoplasia, ductal carcinoma in situ, Helicobacter pylori infections of the stomach, colon polyps, severe hepatitis or cirrhosis (especially virally-induced hepatitis) of the liver, and other premalignant conditions that can progress to cancer.
  • Mesangial cell proliferative disorders refer to disorders brought about by abnormal proliferation of mesangial cells.
  • Mesangial cell proliferative disorders include renal diseases, such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic microangiopathy syndromes and glomerulopathies.
  • Fibrotic disorders are diseases or conditions causing, caused by, involving or exacerbated by the abnormal formation of extracellular matrices, unwanted or excessive fibrosis, or both. Fibrotic disorders can occur in, for instance, skin, liver, kidney, heart or lung tissue.
  • Fibrotic disorders include scarring (e.g., keloid formation and hypertrophic scars), scleroderma, kidney fibrosis (e.g., glomerular sclerosis or renal tubulointerstitial fibrosis), pulmonary fibrosis (including idiopathic pulmonary fibrosis), cardiac fibrosis, chemotherapy/radiation-induced lung fibrosis, pancreatitis, atherosclerosis, restenosis, inflammatory bowel disease, Crohn's disease, arthritis, cancer (e.g., invasive breast cancer, stromal rich mammary tumors, dermatofibromas, angiolipoma and angioleiomyoma), fascitis, general fibrosis syndrome (characterized by replacement of normal muscle tissue by fibrous tissue in varying degrees), liver fibrosis (e.g., hepatic cirrhosis), acute fibrosis (e.g., in response to various forms of trauma, including accidental injuries, infections, surgery, burns, radiation or chemotherapy treatment
  • Hyperproliferative skin disorders include psoriasis, skin cancer and epidermal hyperproliferation.
  • the diketopiperazines of the invention, or a pharmaceutically-acceptable salt or prodrug thereof, can also be used to treat viral infections.
  • Specific viral infections treatable according to the invention include infections caused by hepatitis B virus, hepatitis C virus, rubella virus, human immunodeficiency virus (HIV), human herpesvirus 4 (Epstein-Barr virus), human herpesvirus 5 (human cytomegalovirus or HCMV), human herpesvirus 8 (Kaposi's sarcoma-associated herpesvirus or KSHV), human papillomarvirus (HPV), polyomaviruses, human respiratory syncytial virus (RSV), adenovirus and influenza virus.
  • HIV human immunodeficiency virus
  • human herpesvirus 4 Epstein-Barr virus
  • human herpesvirus 5 human cytomegalovirus or
  • the invention also provides a method of inhibiting the production, release or both of matrix metalloproteinase-9 (MMP-9) by cells.
  • the method comprises contacting the cells with an effective amount of a diketopiperazine of formula I or a pharmaceutically- acceptable salt or prodrug thereof.
  • the cells can be contacted with the diketopiperazine by any method known in the art.
  • the cells can be contacted with the diketopiperazine in vivo by administering an effective amount of the diketopiperazine to an animal.
  • Cells that produce and/or release MMP-9 include those cells found in or around a tumor. Such cells include the tumor cells themselves, stromal cells, eosinophils, macrophages, neutrophils and endothelial cells. See Thiennu H. Vu and Zena Werb, "Gelatinase B: Structure, Regulation, and Function," pages 115-148, in Matrix Metalloproteinases (Academic Press, Editors William C. Parks and Robert P. Mecham, 1998). Cells that produce and/or release MMP-9 also include cells involved in inflammation. See, e.g, Id., Solakivi et al., Lipids in Health and Disease, 8(11) (Epub
  • the invention also provides a method of inhibiting the activation of Akt by cells.
  • the method comprises contacting the cells with an effective amount of a diketopiperazine of formula I or a pharmaceutically-acceptable salt or prodrug thereof.
  • the cells can be contacted with the diketopiperazine by any method known in the art.
  • the cells can be contacted with the diketopiperazine in vivo by administering an effective amount of the diketopiperazine to an animal.
  • Protein kinases are involved in the signal transduction pathways linking growth factors, hormones and other cell regulation molecules to cell growth, survival and metabolism.
  • One such protein kinase is Akt kinase.
  • Akt kinase also known as protein kinase B, is a serine/threonine kinase that plays a central role in promoting the proliferation and survival of a wide range of cell types, thereby protecting cells from apoptosis.
  • a number of protein kinases and phosphatases regulate the activity of Akt. For instance, activation of Akt is mediated by phosphatidylinositol 3'-OH kinase (PI3 kinase or PI3K).
  • PI3 kinase phosphatidylinositol 3'-OH kinase
  • Activated PDK produces phosphatidylinositol-3,4,5-triphosphate (PI(3,4,5)P 3 ) at the inner side of the plasma membrane.
  • the increase in PI(3,4,5)P 3 recruits Akt to the inner membrane, where it is activated.
  • Akt can also be activated by growth signals that are independent of PDK. Full activation of Akt requires phosphorylation at two sites by two different kinases.
  • Activated Akt modulates the function of numerous substrates involved in the regulation of cell proliferation, growth and survival, and of cell cycle entry and progression, and activated Akt is involved in the regulation of numerous cellular processes, including transcription, differentiation, metabolism, apoptosis, migration, metastasis, angiogenesis and fibrosis. As a consequence, Akt plays a role in numerous diseases and conditions.
  • Akt is known to play a critical role in cancer. Activation of Akt contributes to tumorigenesis in many types of tissues, including breast, ovarian, brain, prostate, skin and lymph tissues. Elevated levels of activated Akt have been detected in a variety of cancers, including ovarian, breast, prostate, pancreatic, gastric, colorectal, brain, thyroid, lung, skin, leukemia and undifferentiated tumors (suggesting that Akt may be associated with tumor aggressiveness and progression). Ih addition, it has been found that Akt is constitutively active in a wide array of cancers.
  • the phosphatase PTEN is a critical negative regulator of Akt, and its function is lost in many cancers, including breast and prostate carcinomas, glioblastomas and several cancer syndromes, including Bannayan- Zonana syndrome, Cowden disease and Lhermitte-Duclos disease.
  • Tumor cells without functional PTEN show elevated levels of activated Akt.
  • Cancer treatment by chemotherapy and gamma-irradiation kills target cells primarily by induction of apoptosis, and the anti-apoptotic effects of activated Akt contribute to both chemotherapeutic resistance and radiation resistance.
  • Activated Akt also contributes to tumor invasiveness and metastasis.
  • Akt activation is associated with increased expression and secretion of matrix metalloproteases MMP-9 and MMP-2, and Akt has the ability to up-regulate angiogenesis, both of which also contribute to tumor survival.
  • Akt also plays a role in the life cycle of viruses. See, e.g., Cooray, J. Gen. Virol, 85:1065-1076 (2004) and PCT application WO 2007/149730. In particular, inhibition of apoptosis has become recognized as an important contributory factor in virus survival.
  • PI3K/Akt pathway provides an alternative to the expression of viral oncogenes or the direct inhibition of pro- apoptotic proteins. It has become evident that many viruses require upregulation of this pathway to sustain long-term infections and it is modulated, in some cases, by specific viral products to create an environment favorable for cellular transformation. In other cases, PI3K/Akt signaling simply helps to create an environment favorable for virus replication and virion assembly.
  • Akt plays a role in angiogenesis and fibrosis.
  • Angiogenesis and fibrosis are key components in development, growth, wound healing and regeneration.
  • Activation of Akt is sufficient to induce angiogenesis, and activated Akt plays an important role in several of the processes involved in angiogenesis. See, e.g., Jiang and Liu, Current Cancer Drug Targets, 8:19-26 (2008); Sheng et al., J. Cell. Physiol., 218:451-454 (2009).
  • Activation of the Akt pathway results in the production of connective tissue growth factor (CTGF).
  • CTGF connective tissue growth factor
  • CTGF is a potent growth factor that has been shown to play a role in fibroblast proliferation, cell adhesion and the stimulation of extracellular matrix (ECM) production, and CTGF is a potent activator of fibrosis. Accordingly, Akt is a target of choice for anti- angiogenesis therapy for cancer and other angiogenic diseases and conditions and for treating fibrotic disorders.
  • Akt is the master regulator of the proliferative/migratory response of vessel wall cells to injury.
  • PCT application WO 03/032809. Accordingly, restenosis of vessels after angioplasty and narrowing of implanted blood vessels (such as arteries, veins, vascular grafts and conduits) following implantation can be prevented or reduced by inhibiting Akt activity in the cells of the vessel.
  • the Akt inhibitor is preferably administered locally to the blood vessel (such as through a catheter or by being provided as part of a coating on a stent). Id.
  • Akt activation has also been reported to play a role in inflammation and autoimmunity. See, e.g, Ottonello et al., Br. J. Pharmacol., (March 25, 2009) (Epub ahead of print) (PMID 19338579), Solakivi et al., Lipids in Health and Disease, 8(11) (Epub March 30, 2009), Rane et al., Front. Biosci., 14:2400-2412 (2009), Baker et al., J.
  • Akt-mediated disease or condition is a disease or condition causing, caused by, involving, or exacerbated by, activation of Akt.
  • Akt-mediated diseases and conditions include proliferative diseases and conditions, angiogenic diseases and conditions, cancer, fibrotic disorders, restenosis, viral infections, inflammation and autoimmunity.
  • R 2 is the R 2 side chain of the diketopiperazines of formula I that is primarily responsible for their activity. Further, it is presently believed that optimum activity is obtained when R 2 has formula II, R 5 is aryl, heteroaryl or aryloxy, n is 1 and R 6 is methyl. Preferably, R 5 is aryl or aryloxy. More preferably, R 5 is phenyl or phenoxy, most preferably phenyl. Also, preferably, R 5 is in the 4 (para) position on the ring.
  • Treating is used herein to mean to reduce (wholly or partially) the symptoms, duration or severity of a disease or condition, including curing the disease, or to prevent, or reduce the incidence of, the disease or condition (Le., to cause the symptoms of the disease or condition not to develop in an animal that may be exposed or predisposed to the disease or condition, but does not yet experience or display symptoms of the disease or condition).
  • an effective amount is meant the amount of a compound that, when administered to an animal for treating a disease or condition or for causing an effect is sufficient to do so.
  • the “effective amount” can and will most likely vary depending on the compound, the disease or condition and its severity, or the effect sought to be caused, and the age, weight, etc., of the animal to be treated (see below).
  • a diketopiperazine of formula I or a pharmaceutically- acceptable salt or prodrug thereof, is administered to the animal.
  • the animal is a mammal, such as a rabbit, goat, dog, cat, horse or human.
  • Effective dosage forms, modes of administration and dosage amounts for the compounds of the invention may be determined empirically, and making such determinations is within the skill of the art.
  • the dosage amount will vary with the particular compound(s) employed, the disease or condition to be treated, the severity of the disease or condition, the route(s) of administration, the rate of excretion of the compound, the duration of the treatment, the identity of any other drugs being administered to the animal, the age, size and species of the animal, and like factors known in the medical and veterinary arts.
  • a suitable daily dose of a compound of the present invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect.
  • the daily dosage will be determined by an attending physician or veterinarian within the scope of sound medical judgment.
  • the effective daily dose may be administered as two, three, four, five, six or more sub-doses, administered separately at appropriate intervals throughout the day. Administration of the compound should be continued until an acceptable response is achieved.
  • the compounds of the present invention may be administered to an animal patient for therapy by any suitable route of administration, including orally, nasally, rectally, vaginally, parenterally (e.g., intravenously, intraspinally, intraperitoneally, subcutaneously, or intramuscularly), intracisternally, transdermally, intracranially, intracerebrally, and topically (including buccally and sublingually).
  • suitable routes of administration including orally, nasally, rectally, vaginally, parenterally (e.g., intravenously, intraspinally, intraperitoneally, subcutaneously, or intramuscularly), intracisternally, transdermally, intracranially, intracerebrally, and topically (including buccally and sublingually).
  • the preferred routes of administration are orally and topically.
  • compositions of the invention comprise a compound or compounds of the invention as the active ingredient(s) in admixture with one or more pharmaceutically-acceptable carriers and, optionally, with one or more other drugs.
  • Each carrier must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the animal.
  • Pharmaceutically-acceptable carriers are well known in the art. Regardless of the route of administration selected, the compounds of the present invention are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. See, e.g., Remington's
  • Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, powders, granules or as a solution or a suspension in an aqueous or non-aqueous liquid, or an oil-in-water or water-in-oil liquid emulsions, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia), and the like, each containing a predetermined amount of a compound or compounds of the present invention as an active ingredient.
  • a compound or compounds of the present invention may also be administered as bolus, electuary or paste.
  • the active ingredient i.e., one or more diketopiperazines of formula I and/or pharmaceutically-acceptable salts and/or prodrugs thereof
  • one or more pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents,
  • fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter.
  • compositions may also optionally contain opacifying agents and may be of a composition that releases the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in microencapsulated form.
  • Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3- butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol,
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active ingredient, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of compounds of the invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, drops and inhalants.
  • the active ingredient may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to the active ingredient, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to the active ingredient, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder or mixtures of these substances.
  • Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of compounds of the invention to the body.
  • dosage forms can be made by dissolving, dispersing or otherwise incorporating one or more compounds of the invention in a proper medium, such as an elastomeric matrix material.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate-controlling membrane or dispersing the compound in a polymer matrix or gel.
  • compositions include those suitable for administration by inhalation or insufflation or for nasal or intraocular administration.
  • the compounds of the invention are conveniently delivered from an insufflator, nebulizer or a pressurized pack or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the composition may take the form of a dry powder, for example, a powder mix of one or more compounds of the invention and a suitable powder base, such as lactose or starch.
  • a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form in, for example, capsules or cartridges, or, e.g., gelatin or blister packs from which the powder may be administered with the aid of an inhalator, insufflator or a metered-dose inhaler.
  • compounds of the invention may be administered by means of nose drops or a liquid spray, such as by means of a plastic bottle atomizer or metered-dose inhaler.
  • Liquid sprays are conveniently delivered from pressurized packs. Typical of atomizers are the Mistometer (Wintrop) and Medihaler (Riker).
  • Drops such as eye drops or nose drops, may be formulated with an aqueous or nonaqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents. Drops can be delivered by means of a simple eye dropper-capped bottle or by means of a plastic bottle adapted to deliver liquid contents dropwise by means of a specially shaped closure.
  • compositions of this invention suitable for parenteral administrations comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as wetting agents, emulsifying agents and dispersing agents. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like in the compositions.
  • prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monosterate and gelatin.
  • agents which delay absorption such as aluminum monosterate and gelatin.
  • delayed absorption of a parenterally-administered drug is accomplished by dissolving or suspending the drug in an oil vehicle.
  • injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drag release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drag in liposomes or microemulsions which are compatible with body tissue. The injectable materials can be sterilized for example, by filtration through a bacterial-retaining filter.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • sterile liquid carrier for example water for injection
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the type described above.
  • the diketopiperazines of formula I, or a pharmaceutically-acceptable salt or prodrug thereof may be given alone to treat a disease or condition according to the invention.
  • the diketopiperazines of formula I, or a pharmaceutically- acceptable salt or prodrug thereof may be given in combination with one or more other treatments or drags suitable for treating the disease or condition.
  • the compounds of the present invention can be administered prior to, in conjunction with (including simultaneously with) or after, the other treatment or drag.
  • the compounds of the present invention can be administered prior to, in conjunction with or after, another anti-cancer treatment.
  • anti-cancer treatments include surgery, radiation or chemotherapy using any of a variety of anti-cancer agents.
  • any anti-cancer agent that has activity versus a susceptible tumor being treated may be administered prior to, in conjunction with or after, a diketopiperazine, or a pharmaceutically-acceptable salt or prodrug thereof, in the treatment of cancer according to the present invention.
  • a diketopiperazine or a pharmaceutically-acceptable salt or prodrug thereof
  • Examples of such agents can be found in Devita and Hellman (editors), Cancer Principles and Practice of Oncology, 6 th edition, February 15, 2001, Lippincott Williams & Wilkins (publishers), U.S. Pub. Patent Appl. No. 2008/0009507, PCT Application No. WO 2009/009793 and PCT Application No.
  • Typical anti-cancer agents useful in the present invention include anti-microtubule agents, anti-mitotic agents, platinum coordination complexes, alkylating agents, antibiotic agents, antimetabolites, hormones and hormonal analogs, topoisomerase I inhibitors, topoisomerase II inhibitors, angiogenesis inhibitors, signal transduction pathway inhibitors, proapoptotic agents and immunotherapeutic agents.
  • Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle.
  • examples of such agents include diterpenoids (e.g., paclitaxel and docetaxel) and vinca alkaloids (e.g., vinblastine, vincristine and vinorelbine).
  • Platinum coordination complexes are non-phase specific anti-cancer agents which are interactive with DNA.
  • the platinum complexes enter tumor cells, undergo aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor.
  • Examples of platinum coordination complexes include cisplatin and carboplatin.
  • Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecules such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death.
  • alkylating agents include nitrogen mustards (e.g., cyclophosphamide, melphalan and chlorambucil), alkyl sulfonates (e.g, busulfan), nitrosoureas (e.g, carmustine), triazenes (e.g., dacarbazine) and imidazotetrazines (e.g., temozolomide).
  • nitrogen mustards e.g., cyclophosphamide, melphalan and chlorambucil
  • alkyl sulfonates e.g, busulfan
  • nitrosoureas e.g, carmustine
  • triazenes e.g., dacarbazine
  • imidazotetrazines e.g., temozolomide
  • Antibiotic anti-cancer agents are non-phase specific agents which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death.
  • antibiotic anti-cancer agents include actinomycins (e.g., dactinomycin), anthrocyclins (e.g., daunorubicin and doxorubicin) and bleomycins.
  • Topoisomerase I inhibitors include camptothecin and camptothecin derivatives (e.g, irinotecan, topotecan and the various optical forms of 7-(4-methylpiperazino- methylene)-10,ll-ethylenedioxy-20-camptothecin). Camptothecins' cytotoxic activity is believed to be related to its topoisomerase I inhibitor activity
  • Topoisomerase II inhibitors include epipodophyllotoxins (e.g., etoposide and teniposide). Epipodophyllotoxins are phase specific anti-cancer agents derived from the mandrake plant.
  • Epipodophyllotoxins typically affect cells in the S and G 2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows.
  • Antimetabolite anti-cancer agents are phase specific agents that act at S phase
  • antimetabolite anti-cancer agents include fluorouracil, methotrexate, cytarabine, mercaptopurine, thioguanine, hydroxyurea and gemcitabine.
  • Hormones or hormonal analogs are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth or lack of growth of the cancer.
  • hormones and hormonal analogs useful in cancer treatment include adrenocorticosteroids (e.g., prednisone and prednisolone), aminoglutethimide and other aromatase inhibitors (e.g., anastrozole, letrazole, vorazole and exemestane), progestins (e.g., megestrol acetate), estrogens, androgens and anti-androgens (e.g., flutamide, nilutamide, bicalutamide, cyproterone acetate and 5 ⁇ -reductases, such as finasteride and dutasteride), anti-estrogens (e.g., tamoxifen, toremifene, raloxifene, droloxifene, iod
  • Angiogenesis inhibitors include anti-VEGF antibodies, inhibitors of integrin alpha v beta 3 , endostatin, angiostatin, danazol and those methylphenidate derivatives described in U.S. Pub. Patent Appl. No. 20060189655, particularly ⁇ -[phenyl-4-phenyl]-2- piperidineacetic acid methyl ester and ⁇ -[phenyl-4-phenoxy]-2-piperidineacetic acid methyl ester (i.e., those derivatives of methylphenidate ( ⁇ -phenyl-2-piperidineacetic acid methyl ester) having the hydrogen of the phenyl group at the 4 (para) position replaced with phenyl or phenoxy).
  • Signal transduction pathway inhibitors are those inhibitors which block or inhibit a chemical process which evokes an intracellular change, particularly cell proliferation or differentiation.
  • Signal transduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, serine/threonine kinases, PDKs, myoinositol signaling and Ras oncogenes, and SH2/SH3 domain blockers. Suitable inhibitors are described in PCT Application No. WO 2009/032651 and the references cited therein, and include kinase inhibitors, receptor antagonists, antibodies, ribozymes and anti-sense oligonucleotides.
  • N- ⁇ 3-chloro-4-[(3- fluorobenzyl)oxy]phenyl ⁇ 6-[5-( ⁇ [2-(methanesulfonyl)ethyl]amino ⁇ methyl)-2-furyl]-4- quinazolinamine also known as lapatanib and Tykerb
  • inhibitors of farnesyltransferase include geranyl-geranyl transferase and CAAX proteases, Imclone C225 EGFR specific antibody, Herceptin (trastuzamab), erbB2 antibody, 2CB VEGFR2 specific antibody, BAY-43-9006, CI-1040, PD-098059, Wyeth CCI 779, and LY294002 and cell cycle signaling inhibitors
  • Proapoptotic agents include Genta's G3139 bcl-2 antisense oligonucleotide which downregulates the expressionof bcl-2 in tumors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL).
  • Immunotherapeutic agents include Imclone C225 EGFR specific antibody, Herceptin (trastuzamab), alemtuzumab, Erbitux (cetuximab), Avastin (bevacizumab), gemtuzumab, iodine 131 tositumomab, rituximab, erbB2 antibody, 2CB VEGFR2 specific antibody, anti-VEGF antibodies, and vaccines.
  • Anti-viral drugs include amantadine, rimantadine, pleconaril, lamivudine, fomivirsen, rifampicin, zidovudine, Relenza (zanamivir), Tamiflu (oseltamivir phosphate), Zovirax (acyclovir), interferons and antibodies.
  • Biphenyl-4-yl-(3,6-dioxo-piperazin-2-yl)-acetic acid methyl ester (Cpd. 5) was synthesized as described below and illustrated in Figure 1.
  • Assays were performed to determine the effect of Cpd. 5 (synthesis described in Example 1) on proliferation of two cancer cell lines. These two cell lines were STTG grade 4 astrocytoma cell line and AU565 breast cancer cell line. Both cell lines were obtained from American Type Culture Collection, Rockville, MD (ATCC). To perform the assays, a 50 mM stock solution of Cpd. 5 was prepared in DMSO and frozen at -80 9 C. Aliquots of the stock solution were removed and 100 ⁇ M dilutions were made in Iscove's modified Dulbecco's medium (IMDM; obtained from ATCC) supplemented with 10% fetal bovine serum (FBS) and penicillin/streptomycin solution.
  • IMDM Iscove's modified Dulbecco's medium
  • Controls containing equivalent amounts of DMSO to the 100 ⁇ M solution were mixed for comparative purposes. 100 ⁇ l of the resulting solutions were then added to 96 well culture plates in triplicate and placed in a 37°C incubator with 5% CO 2 to warm.
  • the controls were the PB kinase inhibitor, LY294002, and an inactive PD kinase inhibitor related compound, LY303511 (both obtained from Sigma).
  • Promega cell titer reagent is a solution containing a tetrazolium dye that is reduced by living cells to a formazan dye, and this reduction is proportional to the number of living cells present in the well.
  • the plates were incubated at 37 0 C for an additional 4-18 hours to allow the colorimetric assay to develop, and the OD of each well was determined in a microplate reader using a 530 nm filter. The OD of wells containing cell titer reagent in culture medium with no cells was subtracted from the
  • the % decrease of the total cell titer signal was determined using the following formula. ((Diluent OD - Experimental OD)/ Diluent OD))XlOO. Data are presented in Table 1 as mean % decrease of cell titer signal of three separate experiments, plus or minus standard deviation. In the mean % decrease column, a positive number means a decrease in cell titer signal, while a negative number means an increase in cell titer signal. The p values were calculated versus diluent controls. TABLE 1
  • STTG and AU565 cells were seeded at 10,000 cells/well in 96- well plates and grown until a confluent monolayer was obtained. Under these conditions, the cells should no longer proliferate.
  • the culture medium was IMDM supplemented with 10% FBS. After the cells reached confluence, the culture medium was removed by aspiration and replaced with fresh medium containing the test compounds (see Table 2 below). The cell cultures were incubated for an additional 24 hours in the presence of the added compounds and then the cell titer signal was assayed by adding 20 ⁇ l Promega cell titer aqueous one reagent to each well.
  • the % decrease of the total cell titer signal was determined using the following formula. ((Diluent OD - Experimental OD)/ Diluent OD))XlOO. Data are presented in
  • Table 2 as mean % decrease of cell titer signal of three separate experiments, plus or minus standard deviation, hi the mean % decrease column, a positive number means a decrease in cell titer signal, while a negative number means an increase.
  • the p values were calculated versus diluent controls. This experiment was repeated, except using incubations of 18 hours, instead of 24 hours, in the presence of the compounds and 4 hours, instead of 2 hours, for color development. The results are presented in Table 3 below.
  • MMPs matrix metalloproteinases
  • MMP-9 Matrix metalloproteinase-9
  • Assays were performed to determine the effect of Cpd. 5 (synthesis described in
  • Example 1 on the secretion of MMP-9 by the BTOOl glioma cell line.
  • the BTOOl cell line was established as follows from cells obtained after surgery. The excised tissue was treated briefly with a protease cocktail, and the resulting cell suspension was cultured in EvIDM supplemented with 10% FBS. Expanded cells were then frozen for future use. A fresh 50 mM stock solution of Cpd. 5 was prepared in ethanol and warmed to
  • the BTOOl cells were cultured in 25 cm 2 flasks in IMDM containing 10% FBS at 37 °C and 5% CO 2 . When cells reach 70-80% confluence, they were washed two times with DVIDM that had been warmed to 37 °C in a water bath.
  • the experimental culture media containing Cpd. 5 were then added to the flasks (4.5 ml per flask), and the flasks were incubated at 37 ⁇ C and 5% CO 2 for 48 hours.
  • the conditioned media were then removed from the cells, and cellular debris was removed by centrifugation at 2000 rpm for 10 minutes. The supernatants were then transferred to sterile tubes and stored at -2OX!.
  • Assays were performed to determine the effect of Cpd. 5 on proliferation of an additional cancer cell line.
  • This cell line was the U-118 metastatic astrocytoma cell line. It was obtained from ATCC.
  • a 50 mM stock solution of Cpd. 5 was prepared in DMSO and frozen at -80 °C. Aliquots of the stock solution were removed and dilutions containing from 50 ⁇ M to 300 ⁇ M Cpd. 5 were made in Iscove's modified Dulbecco's medium (IMDM; obtained from ATCC) supplemented with 10% fetal bovine serum (P 7 BS) and penicillin/streptomycin solution. Controls containing equivalent amounts of DMSO to the 300 ⁇ M solution were mixed for comparative purposes. 100 ⁇ l of the resulting solutions were then added to 96 well culture plates in triplicate and placed in a 37°C incubator with 5% CO 2 to warm. The controls included the PI3 kinase inhibitor, LY294002, and an inactive PD kinase inhibitor related compound, LY303511 (both obtained from Sigma).
  • IMDM Iscove's modified Dulbecco's medium
  • P 7 BS fetal bovine serum
  • Cells were removed from passage flasks using trypsin and ethylenediaminetetracetic acid (EDTA) and counted using trypan blue to establish cell counts/ml and viability. For all experiments, viability of cells was greater than 95%. Solutions of 4,000 cells/100 ⁇ l were prepared in the medium from above, and 100 ⁇ l of the solutions were added to each well of the plates. The plates were placed back in the incubator and incubated with the compounds for 96 hours. Following incubation, cell proliferation was assayed by adding 20 ⁇ l Promega cell titer aqueous one reagent to each well.
  • EDTA ethylenediaminetetracetic acid
  • Promega cell titer reagent is a solution containing a tetrazolium dye that is reduced by living cells to a formazan dye, and this reduction is proportional to the number of living cells present in the well.
  • the plates were incubated at 37°C for an additional 4 hours to allow the colorimetric assay to develop, and the OD of each well was determined in a microplate reader using a 530 nm filter. The OD of wells containing cell titer reagent in culture medium with no cells was subtracted from the OD of all experimental readings. The results are presented in Table 5 below.
  • the % decrease of the total cell titer signal was determined using the following formula. ((Diluent OD - Experimental OD)/ Diluent OD))XlOO. Data are presented in Table 5 as mean % decrease of cell titer signal of three separate experiments, plus or minus standard deviation. In the mean % decrease column, a positive number means a decrease in cell titer signal, while a negative number means an increase in cell titer signal. The p values were calculated versus diluent controls.
  • Assays were performed to determine the effect of Cpd. 5 on proliferation of human umbilical vein endothelial cells (HUVECs). Passage 4-5 HUVECs, human source lot number 13047 (obtained from Lonza) were put into the wells of a 96- well tissue culture plate at 1200 cells/well in 100 ⁇ l endothelial growth medium-2 (EGM-2) complete medium (obtained from Lonza). A 50 mM stock solution of Cpd. 5 was prepared in
  • DMSO and frozen at -80 0 C Aliquots of the stock solution were removed and dilutions containing from 50 ⁇ M to 300 ⁇ M Cpd. 5 were made in EGM-2 complete medium (Lonza). Controls containing equivalent amounts of DMSO to the 200 ⁇ M solution were mixed for comparative purposes. 100 ⁇ l of the resulting solutions were then added to the cells in the 96-well culture plates in triplicate and placed in a 37°C incubator with 5% CO 2 for 72 hours. The controls were the PI3 kinase inhibitor, LY294002, and an inactive PD kinase inhibitor related compound, LY303511 (both obtained from Sigma).
  • Promega cell titer reagent is a solution containing a tetrazolium dye that is reduced by living cells to a formazan dye, and this reduction is proportional to the number of living cells present in the well.
  • the plates were incubated at 37°C for an additional 4 hours to allow the colorimetric assay to develop, and the OD of each well was determined in a microplate reader using a 530 nm filter. The OD of wells containing cell titer reagent in culture medium with no cells was subtracted from the OD of all experimental readings.
  • the % decrease of the total cell titer signal was determined using the following formula. ((Diluent OD - Experimental OD)/ Diluent OD))XlOO. Data are presented in Table 7 as mean % decrease of cell titer signal of three separate experiments, plus or minus standard deviation. In the mean % decrease column, a positive number means a decrease in cell titer signal, while a negative number means an increase in cell titer signal.
  • Cpd. 5 at 100 ⁇ M final concentration inhibited the proliferation of HUVECs. At lower concentrations, it appeared to stimulate proliferation of HUVECs. The reason for this stimulation is not known, but may be due to the DMSO in which Cpd. 5 is dissolved.
  • Akt protein kinase B, PKB
  • IGF-I insulin, interleukin-3 (IL-3), IL-6, heregulin and VEGF.
  • Akt has three isoforms, and activation of all three isoforms is similar in that phosphorylation of two sites is necessary for full activity.
  • Akt exerts anti-apoptotic effects through phosphorylation of substrates that directly or indirectly regulate apoptosis.
  • Activation of the phosphatidylinositol 3 kinase (PBK)/Akt signaling pathway contributes to tumorigenesis in many types of tissues, including breast, ovarian, brain, prostate and lymph tissues. It has been found that Akt is constitutively active in an array of cancers and contributes to both chemotherapeutic resistance and radiation resistance.
  • PBK phosphatidylinositol 3 kinase
  • AU565 cells were grown in 75 cm 2 flasks using Iscove's modified Dulbecco's medium (IMDM; obtained from Lonza) supplemented with 10% fetal bovine serum (FBS, HyClone) and 1% penicillin/streptomycin solution (Lonza). Cells were removed from the flasks using trypsin/EDTA (Lonza) and counted using trypan blue to establish cell counts. Cells in the above supplemented IMDM medium were added to the wells of a 96-well plate (20,000 cells/well), and the plates were incubated at 37 0 C, 5% CO 2 , for 24 hours.
  • IMDM Iscove's modified Dulbecco's medium
  • FBS fetal bovine serum
  • Lonza penicillin/streptomycin solution
  • Cells in the above supplemented IMDM medium were added to the wells of a 96-well plate (20,000 cells/well), and the plates were incubated at 37
  • the medium was removed and replaced with medium not containing any serum (serum-free HVIDM), and the plates were incubated at 37°C, 5% CO 2 , for an additional 24 hours.
  • test compounds were added to the plates as follows.
  • a 20 mM stock solution of Cpd. 5 was prepared in DMSO and frozen at -80 9 C. Aliquots of the stock solution were removed and dilutions were made in serum-free HVIDM medium. Controls containing equivalent amounts of DMSO (0.05% and 0.25%) were mixed for comparative purposes. The other control was the PI3 kinase inhibitor, LY294002 (obtained from Sigma). 100 ⁇ l of each test compound solution were then added to 96 well culture plates in duplicate, and the plates were placed back into the incubator and incubated at 37°C, 5% CO 2 , for 1 hour.
  • IGF-I insulin-like growth factor-1
  • the CASETM Kit for AKT S473 quantifies the amount of activated (phosphorylated) Akt protein relative to total Akt protein in parallel assays using a conventional ELISA format with colorimetric detection.
  • the Akt phosphorylation site is serine 473 and is recognized by one of the antibodies used in the one of the two parallel assays to provide the measure of activated Akt protein.
  • the other antibody used in the other parallel assay recognizes Akt to provide the measure of total Akt protein. Both primary antibodies are detected using a horseradish peroxidase-labeled secondary antibody. Addition of the manufacturer's Developing Solution for 10 minutes, followed by addition of the manufacturer's Stop Solution, produces the product which can be measured colorimetrically.
  • the results were calculated as follows. The treatment groups containing IGF- I/DMSO were subtracted from their respective IGF-1/DMSO only groups (positive control). This value was divided by the difference between the respective DMSO only group (negative control) and the respective positive control. This value represents the percent inhibition of Akt phosphorylation attributable to the inhibitor alone since the contribution of DMSO is subtracted out.
  • WM-266-4 a metastatic melanoma cell line obtained from ATCC.
  • the WM-266-4 cells were grown in 25 cm 2 flasks using IMDM medium (obtained from Lonza) supplemented with 10% fetal bovine serum (FBS, HyClone) and 1% penicillin/streptomycin solution (Lonza). Cells were removed from the flasks using trypsin/EDTA (Lonza) and counted using trypan blue to establish cell counts.
  • IMDM medium Cells in the above supplemented IMDM medium were added to the wells of a 96- well plate (10,000 cells/well), and the plates were incubated at 37 0 C, 5% CO 2 , for 24 hours. After 24 hours, the medium was removed and replaced with medium not containing any serum (serum-free IMDM), and the plates were incubated at 37 0 C, 5% CO 2 , for an additional 24 hours.
  • test compounds were added to the plates as follows.
  • a 20 mM stock solution of Cpd. 5 was prepared in DMSO and frozen at -80 °C. Aliquots of the stock solution were removed and dilutions were made in serum-free IMDM medium. Final concentrations of Cpd. 5 were 10 ⁇ M and 50 ⁇ M. Controls containing equivalent amounts of DMSO as the two concentrations of Cpd. 5 (0.05% and 0.25%) were mixed for comparative purposes. The other control was the PI3 kinase inhibitor, LY294002 (obtained from Sigma), final concentration of 50 ⁇ M. 100 ⁇ l of each test compound solution were then added to 96 well culture plates in duplicate, and the plates were placed back into the incubator and incubated at 37 0 C, 5% CO 2 , for 1 hour.
  • Akt phosphorylation promoter 100 ⁇ l of serum-free IMDM containing either 0 or 400 ng/ml IGF-I (obtained from Sigma), an Akt phosphorylation promoter, were added, and the plates incubated at 37°C, 5% CO 2 , for 1 more hour. At the end of this time, the cells were fixed immediately with 4% formaldehyde, refrigerated, and the extent of phosphorylation of Akt determined using the Akt Cellular Activation of Signaling ELISA
  • the CASETM Kit for AKT S473 quantifies the amount of activated (phosphorylated) Akt protein relative to total Akt protein.
  • the results were calculated as follows. The treatment groups containing IGF- I/DMSO were subtracted from their respective IGF-1/DMSO only groups (positive control). This value was divided by the difference between the respective DMSO only group (negative control) and the respective positive control. This value represents the percent inhibition of Akt phosphorylation attributable to the inhibitor alone since the contribution of DMSO is subtracted out.

Abstract

L’invention concerne des dicétopipérazines de formule I. L’invention concerne également des compositions pharmaceutiques renferment les dicétopipérazines, ou leurs sels pharmaceutiquement acceptables ou leurs précurseurs, en tant que principe actif. L’invention a également pour objet des traitements thérapeutiques qui utilisent les dicétopipérazines de formule I, y compris l’inhibition d’une maladie ou d’une affection proliférative, l’inhibition d’une angiogenèse, le traitement d’une maladie ou d’une affection angiogénique, le traitement du cancer et d’affections précancéreuses, le traitement d’un trouble fibrotique, le traitement d’une infection virale, le traitement d’une maladie ou d’une affection médiée par Akt, l’inhibition de la production, de la libération ou des deux de la métalloprotéinase matricielle 9 et l’inhibition de l’activation de Akt.
EP09810870A 2008-05-27 2009-05-27 Procédés et composés thérapeutiques Withdrawn EP2300011A4 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5637908P 2008-05-27 2008-05-27
PCT/US2009/045281 WO2009146320A1 (fr) 2008-05-27 2009-05-27 Procédés et composés thérapeutiques

Publications (2)

Publication Number Publication Date
EP2300011A1 true EP2300011A1 (fr) 2011-03-30
EP2300011A4 EP2300011A4 (fr) 2012-06-20

Family

ID=41377553

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09810870A Withdrawn EP2300011A4 (fr) 2008-05-27 2009-05-27 Procédés et composés thérapeutiques

Country Status (4)

Country Link
US (3) US8217047B2 (fr)
EP (1) EP2300011A4 (fr)
JP (2) JP5856843B2 (fr)
WO (1) WO2009146320A1 (fr)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2774959C (fr) 2000-08-04 2016-05-31 Dmi Biosciences, Inc. Methode d'utilisation de dicetopiperazines et composition contenant ces substances
JP2007500747A (ja) 2003-05-15 2007-01-18 ディーエムアイ バイオサイエンシズ インコーポレイテッド T細胞媒介性疾患の処置
JP5544458B2 (ja) * 2005-07-12 2014-07-09 アンピオ ファーマシューティカルズ,インコーポレイテッド 疾病を治療するための医薬製品の製造においてトリロスタンiiiを使用する方法及びトリロスタンiiiを含む医薬製品
JP5856843B2 (ja) 2008-05-27 2016-02-10 アンピオ ファーマシューティカルズ,インコーポレイテッド ジケトピペラジンを用いた医薬組成物
SG2014008171A (en) 2009-06-22 2014-04-28 Ampio Pharmaceuticals Inc Method for treatment of diseases
CA2810844C (fr) 2010-09-07 2017-03-21 Dmi Acquisition Corp. Compositions de diketopiperazine destinees au traitement de syndrome metabolique et de troubles associes
BR112013005432A8 (pt) * 2010-09-07 2017-12-05 Dmi Acquisition Corp tratamento de doenças
CA2850597A1 (fr) * 2011-09-30 2013-04-04 Vanderbilt University Therapies antivirales avec des inhibiteurs de phospholipase d
SG10201608087WA (en) * 2011-10-10 2016-11-29 Ampio Pharmaceuticals Inc Implantable medical devices with increased immune tolerance, and methods for making and implanting
EP2766029B1 (fr) 2011-10-10 2020-03-25 Ampio Pharmaceuticals, Inc. Traitement de maladie dégénérative articulaire
MX355446B (es) 2011-10-28 2018-04-18 Ampio Pharmaceuticals Inc Tratamiento de rinitis.
DE102011117128A1 (de) * 2011-10-28 2013-05-02 Christian-Albrechts-Universität Zu Kiel Verbindungen zur Therapie der Influenza
US9351979B2 (en) * 2012-12-19 2016-05-31 Ampio Pharmaceuticals, Inc. Methods of treatment of diseases
CA2906864A1 (fr) 2013-03-15 2014-09-18 Ampio Pharmaceuticals, Inc. Compositions pour la mobilisation, l'ecotropisme, l'expansion et la differenciation de cellules souches et leurs methodes d'utilisation
KR20170045274A (ko) 2014-08-18 2017-04-26 앰피오 파마슈티컬스 인코퍼레이티드 관절 징후의 치료
CN104940201A (zh) * 2015-06-04 2015-09-30 浙江大学 一种化合物在制备抑制肿瘤侵袭转移药物中的应用
US11389512B2 (en) 2015-06-22 2022-07-19 Ampio Pharmaceuticals, Inc. Use of low molecular weight fractions of human serum albumin in treating diseases
KR101992534B1 (ko) * 2017-09-19 2019-06-24 동원시스템즈 주식회사 방열 시트
KR102140910B1 (ko) * 2019-03-28 2020-08-04 주식회사 노브메타파마 Chp(사이클로-히스프로)를 포함하는 섬유증의 예방, 개선 또는 치료용 조성물

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997048685A1 (fr) * 1996-06-18 1997-12-24 Glaxo Group Limited Inhibiteurs des metalloproteases
US20020052381A1 (en) * 2000-08-04 2002-05-02 David Bar-Or Method of using diketopiperazines and composition containing them
WO2004103304A2 (fr) * 2003-05-15 2004-12-02 Dmi Biosciences, Inc. Traitement de maladies a mediation des lymphocytes t

Family Cites Families (233)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2507631A (en) * 1944-01-19 1950-05-16 Ciba Pharm Prod Inc Pyridine and piperidine compounds and process of making same
US2957880A (en) * 1953-12-23 1960-10-25 Ciba Pharm Prod Inc Process for the conversion of stereoisomers
GB851972A (en) 1957-07-11 1960-10-19 Rhone Poulenc Sa Acid addition salts of esters of substituted piperidyl phenyl methanols and processes for their production
US3941790A (en) * 1967-07-03 1976-03-02 National Research Development Corporation Bis diketopiperazines
BE765030A (fr) 1970-04-02 1971-08-16 Snam Progetti Composes contenant un noyau de dicetopiperazine et leur procedede fabrication
GB1353304A (en) 1971-03-30 1974-05-15 Sagami Chem Res Process for production of diketopiperazine dihydroxamates and intermediate therefor
US4006261A (en) 1973-09-28 1977-02-01 Firmenich S.A. Flavoring with mixtures of theobromine and cyclic dipeptides
US3928330A (en) 1973-12-19 1975-12-23 Ciba Geigy Corp Substituted piperazinedione carboxylic acids and metal salts thereof
GB1459488A (en) * 1974-03-19 1976-12-22 Wyeth John & Brother Ltd Piperazinedione derivatives
JPS5225019Y1 (fr) 1974-12-19 1977-06-07
US4088649A (en) * 1975-07-02 1978-05-09 Firmenich S.A. Process for preparing a diketo-piperazine
JPS5732272Y2 (fr) 1977-09-29 1982-07-15
US4205057A (en) * 1978-01-17 1980-05-27 Government Of The United States Cerebrospinal fluid protein fragments
JPS6236331Y2 (fr) 1979-05-16 1987-09-16
DE2940654A1 (de) * 1979-10-06 1981-04-16 Bayer Ag, 5090 Leverkusen Dimeres keten der 1,2,,-triazol-3-carbonsaeure
US4289759A (en) 1980-06-23 1981-09-15 Ortho Pharmaceutical Corporation Immunoregulatory diketopiperazine compounds
US4331595A (en) * 1980-06-23 1982-05-25 Ortho Pharmaceutical Corporation Immunoregulatory diketopiperazine compounds
JPS5836225U (ja) 1981-08-31 1983-03-09 株式会社東芝 金属製平パレツト
JPS5973574U (ja) 1982-11-10 1984-05-18 三菱自動車工業株式会社 電磁式燃料噴射装置
US4694061A (en) * 1983-10-12 1987-09-15 Ciba-Geigy Corporation Radiation-sensitive polycondensates, processes for their preparation coated material and its use
NZ212051A (en) 1984-05-18 1988-10-28 Univ Australian Immune response suppression; certain epipolythio- dioxopiperazine derivatives and their preparation
US4806538A (en) * 1984-11-02 1989-02-21 Fujisawa Pharmaceutical Co., Ltd. Piperazine compound as PAF-antagonist
JPH0245309Y2 (fr) 1984-12-21 1990-11-30
CS254868B1 (cs) 1985-06-11 1988-02-15 Jiri Vanzura Způsob výroby cyklických dipeptidů
US4771056A (en) * 1985-08-29 1988-09-13 Roman Rozencwaig Method of medical treatment with serotonin antagonists
US4661500A (en) * 1985-08-29 1987-04-28 Roman Rozencwaig Method of medical treatment with serotonin antagonists
US5047401A (en) * 1985-09-09 1991-09-10 Board Of Regents, The University Of Texas System Use of dipeptide alkyl esters to treat GVHD
US4694081A (en) * 1985-09-23 1987-09-15 Monsanto Company Process to prepare 2,5-diketopiperazines
PT83613B (en) 1985-10-28 1988-11-21 Lilly Co Eli Process for the selective chemical removal of a protein amino-terminal residue
JPS63290868A (ja) 1987-05-22 1988-11-28 Fujisawa Pharmaceut Co Ltd ジケトピペラジン誘導体およびその塩類
US5512544A (en) * 1987-09-13 1996-04-30 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising an anticytokine
US5407926A (en) * 1987-12-29 1995-04-18 Alcon Laboratories, Inc. Ophthalmic composition
US4992552A (en) * 1988-08-31 1991-02-12 Eastman Kodak Company Process for preparation of amino acids
US5144073A (en) 1988-08-31 1992-09-01 Hubbs John C Process for preparation of dipeptides
JP2001055340A (ja) 1988-10-31 2001-02-27 Welfide Corp アルブミン製剤の製造方法
US5238938A (en) * 1989-02-10 1993-08-24 Otsuka Pharmaceutical Co., Ltd. Indole derivatives
DE3906952A1 (de) * 1989-03-04 1990-09-06 Boehringer Mannheim Gmbh (3-(c(pfeil abwaerts)1(pfeil abwaerts)(pfeil abwaerts)6(pfeil abwaerts)-c(pfeil abwaerts)1(pfeil abwaerts)(pfeil abwaerts)8(pfeil abwaerts))alkansulfinyl- und sulfonyl-2-methoxymethyl-propyl)-(2-trimethylammonio-ethyl) phosphate, verfahren zu deren herstellung diese verbindungen enthaltende arzneimittel
US5902790A (en) 1995-10-03 1999-05-11 Cytran, Inc. Pharmaceutical angiostatic dipeptide compositions and method of use thereof
DE68922102T2 (de) 1989-10-13 1995-10-19 Phobos Nv Verfahren zur kontinuierlichen Herstellung von hochmolekularen Polyester-Harzen.
DK0478729T3 (da) 1990-03-15 1996-09-16 Nutrasweet Co Fremgangsmåde til fremstilling af aspartam ud fra et diketopiperazin, og hidtil ukendte mellemprodukter og derivater dertil
US6180616B1 (en) * 1990-05-10 2001-01-30 Atsuo F. Fukunaga Use of purine receptor agonists to alleviate or normalize physiopathologically excited sensory nerve function
US6331318B1 (en) 1994-09-30 2001-12-18 Emisphere Technologies Inc. Carbon-substituted diketopiperazine delivery systems
US5578323A (en) 1992-06-15 1996-11-26 Emisphere Technologies, Inc. Proteinoid carriers and methods for preparation and use thereof
US5693338A (en) 1994-09-29 1997-12-02 Emisphere Technologies, Inc. Diketopiperazine-based delivery systems
US6099856A (en) * 1992-06-15 2000-08-08 Emisphere Technologies, Inc. Active agent transport systems
GB9022543D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Antibody production
CA2095539A1 (fr) 1990-11-14 1992-05-15 Robert D. Cook Diagnostic et traitement de la sclerose en plaques
US5776892A (en) * 1990-12-21 1998-07-07 Curative Health Services, Inc. Anti-inflammatory peptides
JPH04234374A (ja) 1990-12-27 1992-08-24 Ajinomoto Co Inc ジケトピペラジン誘導体の製造方法
GB9102997D0 (en) 1991-02-13 1991-03-27 Pfizer Ltd Therapeutic agents
US5543503A (en) * 1991-03-29 1996-08-06 Genentech Inc. Antibodies to human IL-8 type A receptor
US5538993A (en) * 1991-09-12 1996-07-23 Yissum Research Development Company Certain tetrahydrocannabinol-7-oic acid derivatives
JPH07503456A (ja) 1991-10-28 1995-04-13 サイトラン・リミテッド 医薬的ジペプチド組成物およびその使用法
JPH05244982A (ja) * 1991-12-06 1993-09-24 Sumitomo Chem Co Ltd 擬人化b−b10
JPH05163148A (ja) 1991-12-18 1993-06-29 Kanebo Ltd 抗腫瘍剤
GB9200210D0 (en) 1992-01-07 1992-02-26 British Bio Technology Compounds
US5352461A (en) 1992-03-11 1994-10-04 Pharmaceutical Discovery Corporation Self assembling diketopiperazine drug delivery system
JP3176478B2 (ja) 1992-05-29 2001-06-18 いすゞ自動車株式会社 居眠り運転防止装置
US5418218A (en) * 1992-07-10 1995-05-23 The University Of Maryland At Baltimore Histidyl-proline diketopiperazine (cyclo his-pro) a cns-active pharmacologic agent
US5358938A (en) * 1992-07-13 1994-10-25 Cytomed, Inc. Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
US5648486A (en) * 1992-07-13 1997-07-15 Cytomed, Inc. Compounds and methods for the treatment of inflammatory and immune disorders
US5434151A (en) * 1992-08-24 1995-07-18 Cytomed, Inc. Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
US5463083A (en) 1992-07-13 1995-10-31 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
GB9217331D0 (en) 1992-08-14 1992-09-30 Xenova Ltd Pharmaceutical compounds
EP0656004A1 (fr) 1992-08-20 1995-06-07 Cytomed, Inc. Agents a double fonctionnalite anti-inflammatoire et d'immunosuppression
US5728553A (en) * 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
PL177771B1 (pl) * 1992-12-18 2000-01-31 Cell Med Inc Linie komórek hybrydoma i preparaty przeciwciał
ES2068742B1 (es) 1993-02-11 1995-11-16 Uriach & Cia Sa J Nuevos derivados de piridinio.
WO1994020063A2 (fr) 1993-03-04 1994-09-15 Cytoven International N.V. Tryptophane pharmaceutique contenant des compositions dipeptidiques et modes d'utilisation
US6107050A (en) * 1993-05-03 2000-08-22 The United States Of America As Represented By The Department Of Health And Human Services Diagnostic test for alzheimers disease
JPH09503749A (ja) 1993-07-23 1997-04-15 エルエックスアール バイオテクノロジー インコーポレイテッド アポトーシスおよび関連した状態の処置方法
GB9324872D0 (en) 1993-12-03 1994-01-19 Univ Pasteur Pharmaceutical compounds
IL112627A0 (en) * 1994-02-14 1995-05-26 Xenova Ltd Diketopiperazines, their preparation and pharmaceutical or veterinary compositions containing them
JP2921731B2 (ja) 1994-03-10 1999-07-19 日清製油株式会社 有機溶媒または油脂類のゲル化剤
FR2717484A1 (fr) 1994-03-16 1995-09-22 Pf Medicament Nouveaux composés pseudo-bis-peptidiques analogues de la CCK, leur procédé de préparation, leur utilisation à titre de médicament et les compositions pharmaceutiques les comprenant .
WO1995026734A1 (fr) * 1994-04-04 1995-10-12 Freeman William R Emploi de nucleosides de phosphonylemethoxyalkyle pour traiter la pression intra-oculaire elevee
GB9410387D0 (en) * 1994-05-24 1994-07-13 Xenova Ltd Pharmaceutical compounds
US5550132A (en) * 1994-06-22 1996-08-27 University Of North Carolina Hydroxyalkylammonium-pyrimidines or purines and nucleoside derivatives, useful as inhibitors of inflammatory cytokines
AU2871195A (en) 1994-06-23 1996-01-19 Affymax Technologies N.V. Methods for the synthesis of diketopiperazines
US5817751A (en) * 1994-06-23 1998-10-06 Affymax Technologies N.V. Method for synthesis of diketopiperazine and diketomorpholine derivatives
US5750565A (en) * 1995-05-25 1998-05-12 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5703093A (en) 1995-05-31 1997-12-30 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5792776A (en) * 1994-06-27 1998-08-11 Cytomed, Inc., Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
MX9606635A (es) 1994-06-27 1997-12-31 Cytomed Inc Compuestos y metodos para el tratamiento de desordenes cardiovasculares, inflamatorios e inmunes.
US5843950A (en) 1994-07-29 1998-12-01 Nikken Chemicals Co., Ltd. 1,4-dihydropyridine compound and pharmaceutical composition containing the same
US5561115A (en) 1994-08-10 1996-10-01 Bayer Corporation Low temperature albumin fractionation using sodium caprylate as a partitioning agent
JPH10508583A (ja) * 1994-10-05 1998-08-25 カリ ロダー L−フェニルアラニン、チロシン又はトリプトファン、及びでき得ればビタミンb▲下12▼を配合したロフェプラミンを使用する多発性硬化症及びその他の脱髄性状態の治療
US5658955A (en) * 1994-11-01 1997-08-19 Hitzig; Pietr Combined use of dopamine and serotonin agonists in the treatment of immune disorders
ES2087038B1 (es) 1994-11-07 1997-03-16 Uriach & Cia Sa J Nuevas piperidinas con actividad antagonista del paf.
CZ282794A3 (en) 1994-11-16 1996-04-17 Galena Pentapeptidic precursors of biologically active cyclic dipeptides
US5877174A (en) * 1994-12-01 1999-03-02 Toyama Chemical Co., Ltd. 2,3-diketopiperazine derivatives or their salts
JP3634891B2 (ja) 1995-04-06 2005-03-30 株式会社海洋バイオテクノロジー研究所 キチナーゼ阻害物質
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
US6034057A (en) * 1995-07-06 2000-03-07 Zeneca Limited Peptide inhibitors of fibronectine
KR19990028983A (ko) * 1995-07-14 1999-04-15 피터 제이. 코젠스 디-트레오-메틸페니데이트 및 다른 약품으로 이루어진 조성물
US5811241A (en) 1995-09-13 1998-09-22 Cortech, Inc. Method for preparing and identifying N-substitued 1,4-piperazines and N-substituted 1,4-piperazinediones
US5908850A (en) * 1995-12-04 1999-06-01 Celgene Corporation Method of treating attention deficit disorders with d-threo methylphenidate
US5922736A (en) 1995-12-04 1999-07-13 Celegene Corporation Chronic, bolus administration of D-threo methylphenidate
US6486177B2 (en) 1995-12-04 2002-11-26 Celgene Corporation Methods for treatment of cognitive and menopausal disorders with D-threo methylphenidate
US5665714A (en) 1995-12-07 1997-09-09 Clarion Pharmaceuticals Inc. N-substituted glycerophosphoethanolamines
US6060452A (en) * 1996-03-13 2000-05-09 Cytran, Inc. Analogs of L-Glu-L-Trp having pharmacological activity
US6541224B2 (en) * 1996-03-14 2003-04-01 Human Genome Sciences, Inc. Tumor necrosis factor delta polypeptides
NZ331596A (en) 1996-04-03 1999-07-29 Pepresearch A S Non-dendritic backbone peptide carrier
WO1997036888A1 (fr) 1996-04-03 1997-10-09 Merck & Co., Inc. Inhibiteurs de farnesyl-proteine transferase
US5919785A (en) * 1996-04-03 1999-07-06 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
GB2328155B (en) * 1996-04-12 2000-08-02 Peptide Technology Pty Limited Methods of treating immunopathologies using polyunsaturated fattyacids
US5932579A (en) * 1996-06-18 1999-08-03 Affymax Technologies N.V. Collagenase-1 and stromelysin-1 inhibitors, pharmaceutical compositions comprising same and methods of their use
US5985581A (en) 1996-07-25 1999-11-16 The Mclean Hospital Corporation Use of presenilin-1 for diagnosis of alzheimers disease
WO1998009968A1 (fr) 1996-09-09 1998-03-12 Novartis Animal Health Australasia Pty. Limited Dicetopiperazines terpenylees (drimentines)
NZ335544A (en) 1996-10-04 2001-08-31 Neuronz Ltd Use of GPE (tripeptide or dipeptide) in form of Gly-Pro-Glu or Gly-Pro or Pro-Glu as a neuromodulator
RU2125728C1 (ru) 1996-10-10 1999-01-27 Пермская государственная медицинская академия Способ иммунодиагностики рассеянного склероза
EP0835660A1 (fr) 1996-10-14 1998-04-15 Gaston Edmond Filomena Merckx Composition contenant méthylcobalamin pour le traitement de multiple sclérosis et des maladies démyélinisantes
US6441172B1 (en) * 1996-11-07 2002-08-27 Torrey Pines Institute For Molecular Studies Diketodiazacyclic compounds, diazacyclic compounds and combinatorial libraries thereof
US5932112A (en) 1996-11-27 1999-08-03 Browning Transport Management, Inc. Method and apparatus for killing microorganisms in ship ballast water
RU2112242C1 (ru) 1996-12-09 1998-05-27 Пермская государственная медицинская академия Способ диагностики рассеянного склероза
JPH10226615A (ja) 1997-02-18 1998-08-25 Pola Chem Ind Inc アスパラギン酸フェニルアラニン環状ジペプタイド誘導体を含有する組成物
US5859249A (en) * 1997-02-19 1999-01-12 Takasago International Corporation 2-phenyl-2-(2'-piperidinylidene)acetate derivative, process for manufacturing the same, and process for manufacturing optically active 2-phenyl-2-(2'-piperidinyl)acetate derivative by asymmetrically hydrogenating the same
JPH10245315A (ja) 1997-03-05 1998-09-14 Pola Chem Ind Inc シクロジペプタイド誘導体を含有する組成物
US6306909B1 (en) * 1997-03-12 2001-10-23 Queen's University At Kingston Anti-epileptogenic agents
WO1998040748A1 (fr) 1997-03-14 1998-09-17 Neuromark Diagnostic de troubles neurologiques
US6265535B1 (en) * 1997-05-30 2001-07-24 The Trustees Of The University Of Pennsylvania Peptides and peptide analogues designed from binding sites of tumor necrosis factor receptor superfamily and their uses
RU2128840C1 (ru) 1997-06-16 1999-04-10 Пермская государственная медицинская академия Способ диагностики рассеянного склероза
US6222029B1 (en) * 1997-08-01 2001-04-24 Genset 5′ ESTs for secreted proteins expressed in brain
US5834032A (en) 1997-08-11 1998-11-10 Song; Moon K. Compositions and methods for treating diabetes
US6210705B1 (en) * 1997-12-15 2001-04-03 Noven Pharmaceuticals, Nc. Compositions and methods for treatment of attention deficit disorder and attention deficit/hyperactivity disorder with methylphenidate
AU2114199A (en) 1998-01-13 1999-08-02 Trustees Of The University Of Pennsylvania, The Novel dopamine re-uptake inhibitors and methods of synthesizing and using the same
US7202279B1 (en) 1998-02-11 2007-04-10 Georgetown University Cyclic dipeptides and azetidinone compounds and their use in treating CNS injury and neurodegenerative disorders
EP0939124A3 (fr) 1998-02-24 2001-03-21 Smithkline Beecham Plc Séquences de la MBGP1
AU3370099A (en) 1998-03-31 1999-10-18 Mayo Foundation For Medical Education And Research Use of platelet activating factor (paf) inhibitors to inhibit il-5 induced eosinophil activation or degranulation
AU3467099A (en) 1998-04-03 1999-10-25 Cytran Ltd. Methods for production of therapeutic cytokines
CA2326760A1 (fr) 1998-04-03 1999-10-14 Cytran, Ltd. Utilisation de l-glu-l-trp pour le traitement d'une infection par vih
AU767241B2 (en) 1998-09-14 2003-11-06 Qiang Xu Immunosuppressive agents
US6475743B1 (en) 1998-10-02 2002-11-05 Ischemia Technologies, Inc. Marker useful for detection and measurement of free radical damage and method
WO2000020840A1 (fr) 1998-10-02 2000-04-13 Ischemia Technologies, Inc. Epreuves permettant une evaluation rapide d'etats ischemiques et necessaires a cet effet
US6492179B1 (en) 1998-10-02 2002-12-10 Ischemia Techologies, Inc. Test for rapid evaluation of ischemic states and kit
US6461875B1 (en) * 1998-10-02 2002-10-08 Ischemia Technologies, Inc. Test for rapid evaluation of ischemic states and kit
ATE401342T1 (de) 1998-10-02 2008-08-15 Ischemia Tech Inc Verfahren und materialien zum nachweis und zur messung von durch freie radikale verursachten schäden
DE19847690A1 (de) 1998-10-15 2000-04-20 Brahms Diagnostica Gmbh Verfahren und Substanzen für die Diagnose und Therapie von Sepsis und sepsisähnlichen systemischen Infektionen
US7026322B2 (en) 1998-11-12 2006-04-11 Nereus Pharmaceuticals, Inc. Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds
US6358957B1 (en) * 1998-11-12 2002-03-19 Nereus Pharmaceuticals, Inc. Phenylahistin and the phenylahistin analogs, a new class of anti-tumor compounds
AU3074500A (en) 1999-01-20 2000-08-07 Kyowa Hakko Kogyo Co. Ltd. Proteasome inhibitors
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6127385A (en) * 1999-03-04 2000-10-03 Pharmaquest Limited Method of treating depression using l-threo-methylphenidate
US6395752B1 (en) * 1999-03-04 2002-05-28 Pharmaquest Limited Method of treating depression using 1-threo-methylphenidate
EP1166117A2 (fr) 1999-03-19 2002-01-02 Vanderbilt University Methodes et reactifs destines au diagnostic et au traitement de la sclerose en plaques
US6025502A (en) * 1999-03-19 2000-02-15 The Trustees Of The University Of Pennsylvania Enantopselective synthesis of methyl phenidate
UA74781C2 (en) 1999-04-02 2006-02-15 Abbott Lab Antiinflammatory and immumosuppressive compounds inhibiting cell adhesion
US6090780A (en) * 1999-04-07 2000-07-18 Chandon Prasad Histidyl-proline diketopiperazine and method of use
US6689765B2 (en) * 1999-05-04 2004-02-10 Schering Corporation Piperazine derivatives useful as CCR5 antagonists
JP2000327575A (ja) 1999-05-26 2000-11-28 Teika Seiyaku Kk ジケトピペラジン誘導体含有炎症疾患治療剤および新規なジケトピペラジン誘導体
GB9913458D0 (en) 1999-06-09 1999-08-11 Medeva Europ Ltd The therapeutic use of d-threo-methylphenidate
DE19937721A1 (de) 1999-08-10 2001-02-15 Max Planck Gesellschaft Neue Diketopiperazine
WO2001030337A2 (fr) 1999-10-22 2001-05-03 Orbon Corporation Traitement topique de l'hypertension oculaire, du glaucome, de la retinopathie ischemique et de la degenerescence maculaire liee a l'age par une formulation ophtalmique d'antagonistes de la dopamine
JP2003517468A (ja) 1999-11-12 2003-05-27 ワイス セロトニン5−ht1a活性を有する分枝アダマンチルおよびノルアダマチルアリール−およびアラルキルピペラジン
WO2001036351A2 (fr) * 1999-11-19 2001-05-25 Corvas International, Inc. Antagonistes de l'inhibiteur des activateurs du plasminogène
US20050096323A1 (en) * 1999-11-30 2005-05-05 Novoscience Pharma Inc. Diketopiperazine derivatives to inhibit thrombin
AU2001236005A1 (en) 2000-02-29 2001-09-12 Chugai Seiyaku Kabushiki Kaisha Preparations stabilized over long time
KR20010113918A (ko) 2000-03-10 2001-12-28 추후제출 후안부 안과 질환을 치료하고 예방하기 위한 방법 및 조성물
DE10019879A1 (de) 2000-04-20 2001-10-25 Degussa Verfahren zur Herstellung von 2,5-Diketopiperazinen, neue 2,5-Diketopiperazine und deren Verwendung
US7288545B2 (en) 2000-05-09 2007-10-30 Angiorx Corporation Piperazinedione compounds
EP1282609B1 (fr) * 2000-05-09 2008-12-17 AngioRx Corporation Composes de piperazinedione
DE10026998A1 (de) 2000-05-31 2001-12-13 Fresenius Kabi De Gmbh Verfahren zur Herstellung einer kosmetischen Zusammensetzung, die humanes Serum Albumin umfasst, welches aus transgenen nicht-menschlichen Säugern erhalten wurde
US20030153575A1 (en) * 2000-06-08 2003-08-14 Orme Mark W. Tetracyclic diketopiperazine compounds as pdev inhibitors
US7175844B2 (en) * 2000-07-18 2007-02-13 Joslin Diabetes Center, Inc. Methods of modulating fibrosis
CZ20002680A3 (cs) 2000-07-21 2002-03-13 Ev®En Ing. Csc. Kasafírek Cyklické alkylthiopeptidy
CZ20002681A3 (cs) 2000-07-21 2002-03-13 Ev®En Ing. Csc. Kasafírek Cyklické tyrosinové dipeptidy
AU2001284697A1 (en) 2000-08-04 2002-02-18 Dmi Biosciences, Inc. Method of synthesizing diketopiperazines
US20020132793A1 (en) 2000-08-28 2002-09-19 Mel Epstein Use of methylphenidate compounds to enhance memory
WO2005000203A2 (fr) 2001-10-31 2005-01-06 Sention, Inc. Methodes permettant de traiter la deficience cognitive et d'ameliorer la cognition
US20070208087A1 (en) * 2001-11-02 2007-09-06 Sanders Virginia J Compounds, compositions and methods for the treatment of inflammatory diseases
WO2002062797A2 (fr) 2000-12-29 2002-08-15 Celltech R & D, Inc. Utilisations pharmaceutiques et synthese de dicetopiperazines
GB2372740A (en) 2001-01-17 2002-09-04 Xenova Ltd Diketopiperazines
AU2002225219A1 (en) 2001-01-26 2002-08-06 Oxford Glycosciences (Uk) Ltd Diagnosis and treatment of multiple sclerosis
WO2002083667A2 (fr) 2001-04-13 2002-10-24 Vertex Pharmaceuticals Incorporated Inhibiteurs de c-jun n-terminal kinases (jnk) et d'autres proteines kinases
AU2002305450A1 (en) 2001-05-08 2002-11-18 Yale University Proteomimetic compounds and methods
US7368421B2 (en) * 2001-06-27 2008-05-06 Probiodrug Ag Use of dipeptidyl peptidase IV inhibitors in the treatment of multiple sclerosis
WO2003032809A2 (fr) 2001-10-15 2003-04-24 The Medstar Research Institute Modulation de la reponse dependant de akt permettant de prevenir la restenose
WO2003037247A2 (fr) 2001-10-29 2003-05-08 Yeda Research And Development Co. Ltd. Procedes et compositions pharmaceutiques de modulation dopaminergique de l'adherence et de l'activite de cellules t
US20040063654A1 (en) * 2001-11-02 2004-04-01 Davis Mark E. Methods and compositions for therapeutic use of RNA interference
EP1575976A4 (fr) * 2001-11-02 2006-08-23 Insert Therapeutics Inc Procedes et compositions permettant l'utilisation therapeutique de l'interference arn
GB0128108D0 (en) 2001-11-23 2002-01-16 Astrazeneca Ab Therapeutic use
JP4301003B2 (ja) 2001-12-27 2009-07-22 味の素株式会社 グルタミン酸誘導体の製造方法
WO2003059306A1 (fr) * 2002-01-18 2003-07-24 Unilever Plc Compositions cosmetiques contenant un compose cyclodipeptidique
CA2480832A1 (fr) * 2002-04-05 2003-10-23 Nitromed, Inc. Donneurs de monoxyde d'azote, compositions et procedes d'utilisation, applications correspondantes
ES2204294B2 (es) 2002-07-02 2005-02-01 Universidade De Santiago De Compostela Nuevos antibioticos activos frente al vibrio anguillarum y sus aplicaciones en cultivos de peces, crustaceos, moluscos y otras actividades de acuicultura.
US20040038865A1 (en) * 2002-08-01 2004-02-26 Mannkind Corporation Cell transport compositions and uses thereof
US20080260838A1 (en) 2003-08-01 2008-10-23 Mannkind Corporation Glucagon-like peptide 1 (glp-1) pharmaceutical formulations
US7919497B2 (en) 2002-08-02 2011-04-05 Nereus Pharmaceuticals, Inc. Analogs of dehydrophenylahistins and their therapeutic use
DE60316688T2 (de) 2002-08-02 2008-07-17 Nereus Pharmaceuticals, Inc., San Diego Dehydrophenylahistine und analoge davon sowie ein verfahren zur herstellung von dehydrophenylahistinen und analogen davon
KR20050042146A (ko) * 2002-08-06 2005-05-04 아플라겐 게엠베하 결합 분자
DE10238144A1 (de) 2002-08-15 2004-02-26 Basf Ag Verwendung von Diketopiperazin-Derivaten als photostabile UV-Filter in kosmetischen und pharmazeutischen Zubereitungen
WO2004022569A1 (fr) * 2002-09-03 2004-03-18 Georgetown University Inhibiteurs des akt, compositions pharmaceutiques, et utilisations associees
CA2500652A1 (fr) 2002-10-02 2004-04-15 Dmi Biosciences, Inc. Diagnostic et controle de maladies
US7196169B2 (en) 2002-10-11 2007-03-27 Queen's University At Kingston Isolated post-translationally modified mammalian proteins for monitoring and diagnosing muscle damage
US7354574B2 (en) 2002-11-07 2008-04-08 Advanced Ocular Systems Limited Treatment of ocular disease
AU2003281978A1 (en) 2002-11-22 2004-06-18 Boehringer Ingelheim International Gmbh 2,5-diketopiperazines for the treatment of obesity
US20040137068A1 (en) 2002-12-20 2004-07-15 Rajiv Bhushan Ophthalmic formulation for the prevention and treatment of adverse ocular conditions, particularly those associated with the aging eye
SG145701A1 (en) 2003-07-30 2008-09-29 Xenon Pharmaceuticals Inc Piperazine derivatives and their use as therapeutic agents
HUE028528T2 (en) * 2003-09-03 2016-12-28 Neuren Pharmaceuticals Ltd Neuroprotective bicyclic compounds and pharmaceutical compositions containing them
US20050101582A1 (en) * 2003-11-12 2005-05-12 Allergan, Inc. Compositions and methods for treating a posterior segment of an eye
US20050192290A1 (en) * 2004-01-13 2005-09-01 Isaac Melamed Treatment of behavioral disorders
JP2008505084A (ja) 2004-06-29 2008-02-21 アムゲン インコーポレイティッド フラノピリミジン
JP4970260B2 (ja) 2004-08-20 2012-07-04 プロメティック バイオサイエンシズ,リミテッド 親和性クロマトグラフィーによるタンパク質の逐次的単離および精製スキーム
PL1791542T3 (pl) 2004-08-23 2015-11-30 Mannkind Corp Sole diketopiperazyny do dostarczania leków
US20060189655A1 (en) * 2005-01-20 2006-08-24 David Bar-Or Methylphenidate derivatives and uses of them
CN101123965B (zh) * 2005-01-20 2012-08-22 分子药物研究所公司 哌甲酯衍生物及其应用
CN101115479A (zh) * 2005-02-14 2008-01-30 默克公司 Akt活性抑制剂
US8241656B2 (en) 2005-09-21 2012-08-14 Surmodics, Inc Articles including natural biodegradable polysaccharides and uses thereof
EP2497484A3 (fr) 2006-02-22 2012-11-07 MannKind Corporation Procédé pour améliorer les propriétés pharmaceutiques de microparticules comprenant de la dicétopipérazine et un agent actif
KR101438839B1 (ko) 2006-04-14 2014-10-02 맨카인드 코포레이션 글루카곤 유사 펩타이드 1 (glp-1) 약제학적 제제
US20080017576A1 (en) 2006-06-15 2008-01-24 Rensselaer Polytechnic Institute Global model for optimizing crossflow microfiltration and ultrafiltration processes
AR061240A1 (es) 2006-06-20 2008-08-13 Lilly Co Eli Compuestos de de 4-isoquinolin-fenol,composiciones farmaceuticas que los contienen y su uso como agentes antineoplasicos y/o antivirales.
CA2657578A1 (fr) 2006-07-11 2008-01-17 Harkness Pharmaceuticals, Inc. Procedes de traitement de l'obesite en utilisant des facteurs de satiete
US8231929B2 (en) 2006-11-09 2012-07-31 Cook Medical Technologies Llc Medical device coating process
AU2008274956B2 (en) 2007-07-12 2014-08-21 University Of South Florida Inhibitors of Akt/PKB with anti-tumor activity
US20090038416A1 (en) 2007-08-07 2009-02-12 Aleta Behrman Bonner System and method for biological sample collection and analyte detection
WO2009032651A1 (fr) 2007-08-31 2009-03-12 Smithkline Beecham Corporation INHIBITEURS DE L'ACTIVITÉ Akt
KR20100087291A (ko) 2007-09-25 2010-08-04 아보트 러보러터리즈 케모카인 수용체 길항제로서의 옥타하이드로펜탈렌 화합물
US20090163936A1 (en) 2007-12-21 2009-06-25 Chunlin Yang Coated Tissue Engineering Scaffold
US20100042206A1 (en) 2008-03-04 2010-02-18 Icon Medical Corp. Bioabsorbable coatings for medical devices
US8383124B2 (en) 2008-03-12 2013-02-26 Children's Hospital Medical Center Mobilization of hematopoietic stem cells
JP5856843B2 (ja) 2008-05-27 2016-02-10 アンピオ ファーマシューティカルズ,インコーポレイテッド ジケトピペラジンを用いた医薬組成物
US8314106B2 (en) 2008-12-29 2012-11-20 Mannkind Corporation Substituted diketopiperazine analogs for use as drug delivery agents
CN102741280B (zh) 2009-10-30 2015-12-02 诺维信生物制药丹麦公司 白蛋白变体
BR112013005432A8 (pt) 2010-09-07 2017-12-05 Dmi Acquisition Corp tratamento de doenças
CA2810844C (fr) 2010-09-07 2017-03-21 Dmi Acquisition Corp. Compositions de diketopiperazine destinees au traitement de syndrome metabolique et de troubles associes
WO2012174472A1 (fr) 2011-06-17 2012-12-20 Mannkind Corporation Microparticules de dicétopipérazine de capacité élevée
EP2766029B1 (fr) 2011-10-10 2020-03-25 Ampio Pharmaceuticals, Inc. Traitement de maladie dégénérative articulaire
SG10201608087WA (en) 2011-10-10 2016-11-29 Ampio Pharmaceuticals Inc Implantable medical devices with increased immune tolerance, and methods for making and implanting
MX355446B (es) 2011-10-28 2018-04-18 Ampio Pharmaceuticals Inc Tratamiento de rinitis.
WO2014121210A1 (fr) 2013-02-01 2014-08-07 Ampio Pharmaceuticals, Inc. Procédés de production de dicétopipérazines et compositions en contenant
CA2906864A1 (fr) 2013-03-15 2014-09-18 Ampio Pharmaceuticals, Inc. Compositions pour la mobilisation, l'ecotropisme, l'expansion et la differenciation de cellules souches et leurs methodes d'utilisation
KR20170045274A (ko) 2014-08-18 2017-04-26 앰피오 파마슈티컬스 인코퍼레이티드 관절 징후의 치료

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997048685A1 (fr) * 1996-06-18 1997-12-24 Glaxo Group Limited Inhibiteurs des metalloproteases
US20020052381A1 (en) * 2000-08-04 2002-05-02 David Bar-Or Method of using diketopiperazines and composition containing them
WO2004103304A2 (fr) * 2003-05-15 2004-12-02 Dmi Biosciences, Inc. Traitement de maladies a mediation des lymphocytes t

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO2009146320A1 *

Also Published As

Publication number Publication date
US9522893B2 (en) 2016-12-20
JP2015120765A (ja) 2015-07-02
US20150051224A1 (en) 2015-02-19
JP5856843B2 (ja) 2016-02-10
WO2009146320A1 (fr) 2009-12-03
US8217047B2 (en) 2012-07-10
EP2300011A4 (fr) 2012-06-20
US20120157473A1 (en) 2012-06-21
US20100105698A1 (en) 2010-04-29
US8871772B2 (en) 2014-10-28
JP2011521956A (ja) 2011-07-28

Similar Documents

Publication Publication Date Title
US9522893B2 (en) Therapeutic methods and compounds
KR102515507B1 (ko) 알파 v 인테그린 억제제로서의 피롤 아미드
US8440679B2 (en) Bicyclic compounds and their uses as dual c-SRC / JAK inhibitors
RU2580320C2 (ru) Замещенные бензоазепины в качестве модуляторов toll-подобного рецептора
JP2015533782A (ja) スルファモイル−アリールアミド及びb型肝炎の治療のための薬剤としてのその使用
HRP970031A2 (en) Metalloproteinase inhibitors, pharmaceutical compositions containing them and their pharmaceutical uses, and methods and intermediates useful for their preparation
MX2013007541A (es) Compuestos y composiciones terapeuticas.
AU2011284397A1 (en) Matrix metalloproteinase inhibitors
EA014918B1 (ru) Замещенные бициклические пиримидоновые производные
EP3707142B1 (fr) Dérivés de pyrrolopyrazine à utiliser en tant qu'inhibiteurs de l'intégrine alpha v
KR20090050081A (ko) 응고 인자 IXa 억제제로서 사용하기 위한 이소세린 유도체
JP2008534534A (ja) Mmp阻害剤としての置換テトラヒドロイソキノリン、関連する製造法及び医薬としての使用
CN103102352B (zh) 酪氨酸激酶抑制剂吲哚满酮衍生物
CN104418842A (zh) 取代的吲哚化合物及其使用方法和用途
CN103130775B (zh) 作为酪氨酸激酶抑制剂的吲哚满酮衍生物
EP1917012B1 (fr) Composés modulant la migration cellulaire
CN108752241B (zh) 用于治疗关节病的酰基胍类
CN104530029A (zh) 作为Xa因子抑制剂的杂环化合物及其使用方法和用途
CN103848814A (zh) 作为酪氨酸激酶抑制剂的取代吲哚满酮衍生物
CN104496838B (zh) 取代环丁烷类神经氨酸酶抑制剂及其使用方法和用途
CN104497008A (zh) 取代噁唑烷酮类化合物及其使用方法和用途
CN104447390B (zh) 取代环丁烷类神经氨酸酶抑制剂及其使用方法和用途
CN104496839A (zh) 取代环丁烷类神经氨酸酶抑制剂及其使用方法和用途
CN117957228A (zh) sGC刺激剂
AU2010226618A1 (en) Improved chemical synthesis of diazaindoles by Chichibabin cyclization

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20101223

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20120518

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 241/08 20060101AFI20120511BHEP

Ipc: A61K 31/497 20060101ALI20120511BHEP

Ipc: A61P 19/02 20060101ALI20120511BHEP

Ipc: A61P 35/00 20060101ALI20120511BHEP

17Q First examination report despatched

Effective date: 20130708

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: AMPIO PHARMACEUTICALS, INC.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: AMPIO PHARMACEUTICALS, INC.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20180320

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20181201